1
|
Deng W, Xu Z, Hua T, Ji G, Wang Z, Liu P, Zhang Y, Li S, Chao Y, Qian M, Zhao Q, Tian J. Targeted codelivery of nitric oxide and hydrogen sulfide for enhanced antithrombosis efficacy. Bioact Mater 2025; 48:29-42. [PMID: 40303966 PMCID: PMC12038452 DOI: 10.1016/j.bioactmat.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/25/2025] [Accepted: 02/06/2025] [Indexed: 05/02/2025] Open
Abstract
Thrombosis is a leading cause of mortality worldwide. As important gaseous signaling molecules, both nitric oxide (NO) and hydrogen sulfide (H2S) demonstrate antiplatelet and anticoagulant functions, but little attention has been given to their synergistic effect and the underlying mechanism. In the present study, we developed an NO/H2S codelivery system based on enzyme prodrug therapy (EPT) strategy in which the prodrugs are specifically recognized by the engineered β-galactosidase. Targeted codelivery of NO and H2S in vivo was demonstrated by near-infrared fluorescence imaging and confirmed by measuring plasma and tissue levels; as a result, the side effects caused by systemic delivery, such as bleeding time, were reduced. Delivery of an optimized combination of NO and H2S with a low combination index (CI) results in a synergistic effect on the inhibition of platelet adhesion and activation. Mechanistically, NO and H2S cooperatively enhance the cGMP level through redox-based posttranslational modifications of phosphodiesterase 5A (PDE5A), which leads to activation of the cGMP/PKG signaling pathway. Furthermore, targeted codelivery of NO and H2S demonstrates enhanced therapeutic efficacy for thrombosis in two mouse models of FeCl3-induced arterial thrombosis and deep vein thrombosis. Collectively, these results confirm the synergistic efficacy of NO and H2S for antithrombotic therapy, and the codelivery system developed in this study represents a promising candidate for clinical translation.
Collapse
Affiliation(s)
- Weiliang Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zhixin Xu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Tong Hua
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Guangbo Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Zihang Wang
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, 150086, China
| | - Pei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yupeng Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Shuo Li
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, 150086, China
| | - Yuqiu Chao
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, 150086, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, 300071, China
- The Institute of Cardiovascular Sciences, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Health Science Center, Peking University, Beijing, 100191, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Provincial Key Laboratory of Panvascular Disease, Harbin, 150086, China
| |
Collapse
|
2
|
Fu L, Han K, Qi J, Lei Q, Wang X, Wu J, Yang N, Li Y, Kang Y, Yu X, Zhang L, Mao M, He J. Electrohydrodynamically-printed Serpentine Fiber Scaffolds with Enhanced Conductivity and Elasticity for Post-Myocardial Infarction Repair. Acta Biomater 2025:S1742-7061(25)00339-3. [PMID: 40339971 DOI: 10.1016/j.actbio.2025.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/21/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Myocardial infarction remains a leading threat to cardiovascular health, with electrical conduction abnormalities in the infarcted myocardium significantly exacerbating cardiac dysfunction. Enhancing the conductive microenvironment in the infarcted region is essential for promoting myocardial repair. In this study, we developed gold-coated serpentine microfiber-based cardiac scaffolds using electrohydrodynamic printing, which mimicked the intricate architecture of the myocardial tissue's fibrous membrane and allowed for up to 20 % elastic deformation, similar to the maximum strain of the natural heart. Compared to uncoated serpentine scaffolds and traditional linear cardiac scaffolds, the gold-coated serpentine cardiac scaffolds demonstrated enhanced expression of myocardial-specific proteins, including connexin-43 and α-actinin, increased myocardial cell contraction frequency, and better mechanical compatibility with natural cardiac deformation. In a rat model of myocardial infarction, implantation of gold-coated serpentine cardiac scaffolds over four weeks provided significant mechanical support to the infarcted region, reduced myocardial hypertrophy, and markedly improved left ventricular remodeling and cardiac function. Collectively, our findings highlight the potential of serpentine conductive fiber scaffolds as a promising therapeutic strategy for post-myocardial infarction repair, offering innovative insights into the treatment of heart diseases. STATEMENT OF SIGNIFICANCE: This study introduces gold-coated serpentine microfiber scaffolds, created via electrohydrodynamic printing, as a promising solution for post-myocardial infarction repair. These scaffolds, designed to mimic the natural myocardial architecture, offer up to 20 % elastic deformation and enhanced electrical conductivity. Their superior mechanical properties, biocompatibility, and ability to support myocardial cell function make them a promising strategy for restoring heart function post-infarction.
Collapse
Affiliation(s)
- Liyan Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China; State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China
| | - Kang Han
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; State Industry-Education Integration Center for Medical Innovations; Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China
| | - Jie Qi
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Qi Lei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan Shanxi 030024, PR China
| | - Xiaomin Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Jinmin Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Na Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Ying Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Yuming Kang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China
| | - Xiaojing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center; Institute of Cardiovascular Sciences, Translational Medicine Institute, Key Laboratory of Environment and Genes Related to Diseases, Xi'an Jiaotong University, Ministry of Education; Xi'an Shaanxi 710061, PR China.
| | - Liuyang Zhang
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China.
| | - Mao Mao
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; State Industry-Education Integration Center for Medical Innovations; Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China.
| | - Jiankang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Additive Manufacturing Medical Devices, Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China; State Industry-Education Integration Center for Medical Innovations; Xi'an Jiaotong University, Xi'an Shaanxi 710049, PR China.
| |
Collapse
|
3
|
Jiang W, Guo Z, Wang Q, Chen Z, Dong W, Liang Q, Hao Y, Pan H, Zeng C, Liu H, Wang Y. Enhanced nanoparticle delivery across vascular basement membranes of tumours using nitric oxide. Nat Biomed Eng 2025:10.1038/s41551-025-01385-w. [PMID: 40316687 DOI: 10.1038/s41551-025-01385-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 04/03/2025] [Indexed: 05/04/2025]
Abstract
The delivery of nanoparticles (NPs) into solid tumours is challenged by the tumour vascular basement membrane (BM), a critical barrier beneath the endothelium with robust mechanical properties resistant to conventional treatments. Here we propose an approach that uses nitric oxide (NO) to induce the opening of endothelial junctions, creating gaps between endothelial cells and enabling the navigation of NPs through these gaps. Subsequently, NO orchestrates a transient degradation of the BM encasing NP pools in a precise, localized action, allowing the enhanced passage of NPs into the tumour interstitial space through explosive eruptions. We have engineered a NO nanogenerator tailored for near-infrared laser-triggered on-demand NO release at tumour sites. Through breaching the BM barrier, this system results in an increase of clinical nanomedicines within the tumour, boosting the tumour suppression efficacy in both mouse and rabbit models. This approach delicately manages BM degradation, avoiding excessive degradation that might facilitate cancer metastasis. Our NO nanogenerator serves as a precise spatial catalytic degradation strategy for breaching the tumour vascular BM barrier, holding promise for NP delivery into non-tumour diseases.
Collapse
Affiliation(s)
- Wei Jiang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Zixuan Guo
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qin Wang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Macau, China.
| | - Ziqi Chen
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Wang Dong
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qirui Liang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Yinghong Hao
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Huimin Pan
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Cici Zeng
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hang Liu
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Anhui Province Engineering Research Center of Critical Electronic Materials, School of Chemistry and Chemical Engineering, Anhui University, Hefei, China.
| | - Yucai Wang
- National Key Laboratory of Immune Response and Immunotherapy, Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China.
- School of Biomedical Engineering, Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, China.
| |
Collapse
|
4
|
Lau C, Primus CP, Shabbir A, Chhetri I, Ono M, Masucci M, Bin Noorany Aubdool MA, Amarin J, Hamers AJ, Khan Z, Kumar NA, Montalvo Moreira SA, Nuredini G, Osman M, Whitear C, Godec T, Kapil V, Massimo G, Khambata RS, Rathod KS, Ahluwalia A. Accelerating inflammatory resolution in humans to improve endothelial function and vascular health: Targeting the non-canonical pathway for NO. Redox Biol 2025; 82:103592. [PMID: 40209616 PMCID: PMC12005330 DOI: 10.1016/j.redox.2025.103592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Chronic cardiovascular diseases (CVD) are characterised by low-grade systemic inflammation in part due to reduced nitric oxide (NO) bioavailability associated with endothelial dysfunction. Bioavailability of NO can be enhanced by activation of the non-canonical pathway, through increased dietary inorganic nitrate consumption with the potential to attenuate inflammation. METHODS We sought to determine whether dietary inorganic nitrate influences the inflammatory response in models of localised (cantharidin-induced blisters) and systemic inflammation (typhoid vaccine), in healthy male volunteers and conducted two clinical trials; Blister-NITRATE and Typhoid-NITRATE respectively. RESULTS We show that dietary nitrate attenuates endothelial dysfunction following typhoid vaccine administration and accelerates resolution of cantharidin-induced blisters. Both phenomena were associated with an increased level of pro-resolving mediators consequent to a reduction in the expression and activity of pro-inflammatory monocytes. Moreover, we show that leukocytes of the monocyte lineage express the nitrite reductase XOR, that may drive localised nitrite reduction to elevate NO (and cGMP) to drive the protective phenotype. CONCLUSIONS Inorganic nitrate improves endothelial function in the setting of systemic inflammation. Whilst the immediate inflammatory response appeared unaffected by inorganic nitrate treatment, during the resolution phase of the acute inflammatory response lower levels of pro-inflammatory classical inflammatory and intermediate monocytes and attenuated levels of inflammatory cytokines and chemokines were evident. We propose that this reflects a pro-resolution phenotype that may be of potential therapeutic benefit in patients with established CVD. CLINICAL TRIAL REGISTRATION URL: https://www. CLINICALTRIALS gov; unique identifiers NCT02715635, NCT03183830.
Collapse
Affiliation(s)
- Clement Lau
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christopher P Primus
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Asad Shabbir
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Ismita Chhetri
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mutsumi Ono
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michael Masucci
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muhammad Aadil Bin Noorany Aubdool
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Julie Amarin
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alexander Jp Hamers
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Zara Khan
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nitin Ajit Kumar
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Gani Nuredini
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Miski Osman
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Charlotte Whitear
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Tom Godec
- Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Vikas Kapil
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gianmichele Massimo
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rayomand S Khambata
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Krishnaraj S Rathod
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Department of Cardiology, Barts Heart Centre, 2 St. Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - Amrita Ahluwalia
- Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Cardiovascular Clinical Trials Unit, Barts and the London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
5
|
Jalandhra GK, Srethbhakdi L, Davies J, Nguyen CC, Phan PT, Och Z, Ashok A, Lim KS, Phan HP, Do TN, Lovell NH, Rnjak-Kovacina J. Materials Advances in Devices for Heart Disease Interventions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2420114. [PMID: 40244561 DOI: 10.1002/adma.202420114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/07/2025] [Indexed: 04/18/2025]
Abstract
Heart disease encompasses a range of conditions that affect the heart, including coronary artery disease, arrhythmias, congenital heart defects, heart valve disease, and conditions that affect the heart muscle. Intervention strategies can be categorized according to when they are administered and include: 1) Monitoring cardiac function using sensor technology to inform diagnosis and treatment, 2) Managing symptoms by restoring cardiac output, electrophysiology, and hemodynamics, and often serving as bridge-to-recovery or bridge-to-transplantation strategies, and 3) Repairing damaged tissue, including myocardium and heart valves, when management strategies are insufficient. Each intervention approach and technology require specific material properties to function optimally, relying on materials that support their action and interface with the body, with new technologies increasingly depending on advances in materials science and engineering. This review explores material properties and requirements driving innovation in advanced intervention strategies for heart disease and highlights key examples of recent progress in the field driven by advances in materials research.
Collapse
Affiliation(s)
- Gagan K Jalandhra
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lauryn Srethbhakdi
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - James Davies
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Chi Cong Nguyen
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Phuoc Thien Phan
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Zachary Och
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Aditya Ashok
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Khoon S Lim
- School of Medical Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Hoang-Phuong Phan
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thanh Nho Do
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Nigel H Lovell
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Tyree Institute of Health Engineering (IHealthE), University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
- Tyree Institute of Health Engineering (IHealthE), University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
6
|
Zhong X, Luo L, Wu J, Li W, Liu X, Ye T, Li Z, Shi P. Adhesion-Assisted Antioxidant-Engineered Mesenchymal Stromal Cells for Enhanced Cardiac Repair in Myocardial Infarction. ACS NANO 2025; 19:11412-11426. [PMID: 40073336 DOI: 10.1021/acsnano.5c00820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Mesenchymal stromal cell (MSC) therapy holds great promise for treating myocardial infarction (MI). However, the inflammatory and reactive oxygen species (ROS)-rich environment in infarcted myocardium challenges MSC survival, limiting its therapeutic impact. In this study, we demonstrate that chemical modification of MSCs with anti-VCAM1 and polydopamine (PD) significantly enhances MSC survival and promotes cardiac repair. Anti-VCAM1 modification facilitates MSC adhesion to inflamed tissue, ensuring MSC retention in the injured myocardium, while PD scavenges ROS surrounding MSCs, creating a conducive environment for cell transplantation. Our data indicate that chemically engineered MSCs effectively disrupt the inflammation-ROS cycle and modulate inflammation-related immune responses, thus improving MI microenvironments. Single-cell RNA sequencing of rat hearts reveals that treatment with engineered MSCs inhibits cardiac fibrosis by suppressing HB-EGF-EGFR signaling between anti-inflammatory macrophages and activated fibrillates. Ultimately, engineered MSCs demonstrate superior therapeutic efficacy in a rat model of MI. This study presents a straightforward, safe, and efficient chemical method for enhancing MSC therapy.
Collapse
Affiliation(s)
- Xianghua Zhong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Li Luo
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Jiyuan Wu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Weirun Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Xinyang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Tenghui Ye
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
| | - Zhenhua Li
- The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, P. R. China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
7
|
Yang R, Yang L, Zhang N, Wan Y, Chen S, Xiao Y, Liang X, Yang S, Zhong Y, Huang D, Chen W, Zhao B. Targeted delivery of polymeric NO-donor micelles to hepatic stellate cells for restoration of liver function and inhibition of hepatic fibrosis. J Control Release 2025; 379:466-477. [PMID: 39824287 DOI: 10.1016/j.jconrel.2025.01.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Liver fibrosis is a prevalent liver disease associated with significant morbidity, and the activation of hepatic stellate cells (HSCs) serves as the primary causative factor driving the progression of liver fibrosis. However, capillarization of liver sinusoidal endothelial cells (LSECs) induced by hepatic fibrosis can reduce nitric oxide (NO) production and bioavailability, which consequently loses the ability to retain HSCs dormant, leading to amplified HSCs activation. Herein, an elaborate micelle (VN-M@BN) loaded with benazepril (BN) was constructed by self-assembly of polymeric NO donor, aiming for the controlled release of NO in liver fibrosis lesions thereby impeding the progression of liver fibrosis. VN-M@BN with the vitamin A (VA) ligand modification was designed to target HSCs for efficient liver fibrosis inhibition. Controlled NO release significantly downregulated α-smooth muscle actin (α-SMA) and induced apoptosis of activated HSCs, thus enhancing the inhibition effects of BN towards HSCs. Furthermore, the in suit antifibrotic treatment results confirmed that VN-M@BN possessed good circulatory stability and targetability to liver fibrotic tissues, thereby effectively ameliorating the collagen deposition and fibrosis process in damaged liver tissues. The NO-based targeted nanodrug system enabled precise delivery of therapeutic drugs to activated HSCs, thereby synergizing the efficacy in treating liver fibrosis with minimal adverse effects.
Collapse
Affiliation(s)
- Rui Yang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Lifen Yang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Ni Zhang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yuqing Wan
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Shineng Chen
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yiqing Xiao
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoping Liang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Shangjie Yang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China
| | - Dechun Huang
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Wei Chen
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 211198, China.
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
8
|
Zhang W, Peng D, Cheng S, Ni R, Yang M, Cai Y, Chen J, Liu F, Liu Y. Inflammatory Cell-Targeted Delivery Systems for Myocardial Infarction Treatment. Bioengineering (Basel) 2025; 12:205. [PMID: 40001724 PMCID: PMC11852162 DOI: 10.3390/bioengineering12020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Myocardial infarction (MI) is a cardiovascular disease (CVD) with high morbidity and mortality worldwide, which is a serious threat to human life and health. Inflammatory and immune responses are initiated immediately after MI, and unbalanced inflammation post-MI can lead to cardiac dysfunction, scarring, and ventricular remodeling, emphasizing the critical need for an effective inflammation-regulating treatment. With the development of novel therapies, the drug delivery system specific to inflammatory cells offers significant potential. In this review, we introduce immune cells and fibroblasts involved in the development of MI and summarize the newly developed delivery systems related to the use of injectable hydrogels, cardiac patches, nanoparticles, and extracellular vesicles (EVs). Finally, we highlight the recent trends in the use of inflammatory cell-targeting drug delivery systems involving different strategies that facilitate the effective treatment of MI.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yao Liu
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing 400042, China; (W.Z.); (D.P.)
| |
Collapse
|
9
|
Li T, Ding L, Wang Q, Ma J, Wang S. Enhancing cardiac repair post-myocardial infarction: a study on GATM/Gel hydrogel therapeutics. Cell Biol Toxicol 2025; 41:44. [PMID: 39937362 PMCID: PMC11821695 DOI: 10.1007/s10565-025-09987-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND AND PURPOSE Significant advancements in therapeutic approaches are imperative to address the prevalent impact of myocardial infarction (MI) on morbidity and mortality rates worldwide. This study explores the therapeutic potential of GATM/Gel hydrogel, focusing on its ability to enhance cardiac repair and functionality after MI through modulation of inflammatory and repair pathways. EXPERIMENTAL APPROACH The effects of GATM/Gel hydrogel on cardiac recovery were studied in a murine MI model. HA-CHO and gelatin solutions were mixed in situ using a dual syringe with a static mixing needle, and the resulting hydrogel was applied directly to the epicardium during MI modeling, followed by repositioning of the heart and closure of the thorax. Comprehensive in vivo assessments-including echocardiography, electrocardiography, and histopathological analysis-were combined with molecular techniques such as RT-qPCR, Western blotting, and immunofluorescence to elucidate the underlying mechanisms. Key cellular and molecular changes were tracked, focusing on macrophage polarization, angiogenesis, and modulation of the TNF/TNFR2 signaling pathway. KEY RESULTS Employing the GATM/Gel hydrogel led to a substantial improvement in heart function, shown through enhanced ejection fraction and fractional shortening, and reduced infarction size compared to control groups. Mechanistically, the hydrogel promoted the polarization of anti-inflammatory M2 macrophages and stimulated angiogenesis. Moreover, treatment with GATM/Gel hydrogel altered the TNF/TNFR2 pathway, pivotal in mediating inflammatory responses and facilitating myocardial repair. The discoveries highlight the possibility of GATM/Gel hydrogels as an innovative remedy for MI, providing a twofold role in regulating inflammation and fostering recovery.
Collapse
Affiliation(s)
- Te Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lijuan Ding
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Qiang Wang
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Jianing Ma
- Department of Radiation Oncology & Therapy, The First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Shudong Wang
- Department of Cardiology, The First Hospital of Jilin University, No. 1 Xinmin Street Avenue, Chaoyang District, Changchun, 130021, China.
| |
Collapse
|
10
|
Bhullar SK, Thingnam R, Kirshenbaum E, Nematisouldaragh D, Crandall M, Willerth SM, Ramkrishna S, Rabinovich-Nikitin I, Kirshenbaum LA. Living Nanofiber-Enabled Cardiac Patches for Myocardial Injury. JACC Basic Transl Sci 2025; 10:227-240. [PMID: 40131159 PMCID: PMC11897462 DOI: 10.1016/j.jacbts.2024.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 03/26/2025]
Abstract
Because the adult heart has only minimal regenerative capacity, its inability to induce regeneration is well-known in patients with myocardial infarction. However, based on multidisciplinary approaches, it is possible to restore myocardial capability with regenerative medicine via living cardiac patches seeded with therapeutic ingredients ranging from multiple cell types to bioactive molecules, including growth factors, microRNA, and extracellular vesicles to the affected site. Biomaterials, natural and/or synthesized polymers, or in vivo sources such as collagen, fibrin, and decellularized extracellular matrix are used to form these cardiac patches. Herein, we review various techniques where seeded cells and bioactive agents are incorporated within porous nanofibers to create functional cardiac patches that provide myocardial extracellular matrix-like features, mechanical support, and a large surface-to-volume ratio for promoting cellular metabolism as well as compensation for the loss of cardiomyocytes in the infarcted region. We summarize recent advances through electrospinning-generated nanofibers of synthetic and/or natural polymers combined with biological material to create cardiac patches to repair and improve the function of infarcted myocardium. As tailoring designs on cardiac patches have been shown to exhibit deformation mechanisms and enhanced myocardial tissue regeneration, significant roles of various patterns and associated parameters are also discussed. The enhanced delivery of therapeutics offered by tailored nanofiber cardiac patches to treat myocardial infarction and overcome challenges of existing cardiac regeneration therapies such as low stability, short half-lifetime, and delivery methods may promote the potential for their clinical impact on myocardial regeneration.
Collapse
Affiliation(s)
- Sukhwinder K Bhullar
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Raneeta Thingnam
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eryn Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Darya Nematisouldaragh
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Molly Crandall
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Stephanie M Willerth
- Division of Medical Sciences, Centre for Advanced Materials and Technology, University of Victoria, Victoria, British Columbia, Canada
| | - Seeram Ramkrishna
- National University of Singapore, Nanoscience and Nanotechnology Initiative, Engineering Drive, Singapore
| | - Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St Boniface Hospital Albrechtsen Research Centre, Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Pharmacology and Therapeutics, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
11
|
Hou A, Lin Z, Cheng Y, Tang Y, Chen Q, Jiang L, Li L, Zhang Z. A photo-triggered dual-gas donor of nitric oxide and hydrogen sulfide with fluorescence for real-time monitoring of its release. Analyst 2025; 150:378-385. [PMID: 39676677 DOI: 10.1039/d4an01156b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Multi-gaseous signal molecules play a significant role in regulating various physiological and pathological processes. Therefore, studying the synergistic effects of multi-gas donors on biological systems is essential. However, different types of gas donors vary significantly in terms of gas release, including location, dose, and flux. These variations can have a dramatic impact on the biological effects of the gases and require further analysis for confirmation. Thus, a controllable dual-gas donor that enables self-reporting would be valuable for studying the biological effects of two species of gaseous signal molecules. Herein, we present a novel photo-triggered donor, NHD545, which releases nitric oxide and hydrogen sulfide simultaneously. Furthermore, the dual-gas release from this donor is accompanied by turn-on fluorescence, which could be utilized to monitor the NO and H2S release by microscopy. With NHD545, it is convenient to investigate the synergistic effect of dual-gas bioavailability in vitro.
Collapse
Affiliation(s)
- Afeng Hou
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Zhenmei Lin
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Yongfang Cheng
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Yaoping Tang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Qing Chen
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Lingfeng Jiang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Li Li
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
| | - Ziqian Zhang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning 530200, China.
- Guangxi Key Laboratory of Marine Drugs, Nanning 530200, China
| |
Collapse
|
12
|
Wang Z, Li M, Chen J, Zhang S, Wang B, Wang J. Immunomodulatory Hydrogel for Electrostatically Capturing Pro-inflammatory Factors and Chemically Scavenging Reactive Oxygen Species in Chronic Diabetic Wound Remodeling. Adv Healthc Mater 2024; 13:e2402080. [PMID: 39380409 DOI: 10.1002/adhm.202402080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/28/2024] [Indexed: 10/10/2024]
Abstract
Diabetic wound exhibits the complex characteristics involving continuous oxidative stress and excessive expression of pro-inflammatory cytokines to cause a long-term inflammatory microenvironment. The repair healing of chronic diabetic wounding is tremendously hindered due to persistent inflammatory reaction. To address the aforementioned issues, here, a dual-functional hydrogel is designed, consisting of N1-(4-boronobenzyl)-N3-(4-boronophenyl)-N1, N1, N3, N3-tetramethylpropane-1, 3-diaminium (TSPBA) modified polyvinyl alcohol (PVA) and methacrylamide carboxymethyl chitosan (CMCSMA) can not only electrostatically adsorb proinflammatory cytokines of IL1-β and TNF-α, but can also chemically scavenge the excessive reactive oxygen species (ROS) in situ. Both in vitro and in vivo evaluations verify that the negatively charged and ROS-responsive hydrogel (NCRH) can effectively modulate the chronic inflammatory microenvironment of diabetic wounds and significantly enhance wound remodeling. More importantly, the well-designed NCRH shows a superior skin recovery in comparison with the commercial competitor product of wound dressing. Consequently, the current work highlights the need for new strategies to expedite the healing process of diabetic wounds and offers a wound dressing material with immunomodulation.
Collapse
Affiliation(s)
- Zihao Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Mengyu Li
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Jia Chen
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| | - Shengmin Zhang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Bin Wang
- Department of General Surgery, Shenzhen Children's Hospital, Shenzhen, 518038, China
| | - Jianglin Wang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, 518000, China
| |
Collapse
|
13
|
Mohamed ATAE, Ragheb MA, Shehata MR, Mohamed AS. In vivo cardioprotective effect of zinc oxide nanoparticles against doxorubicin-induced myocardial infarction by enhancing the antioxidant system and nitric oxide production. J Trace Elem Med Biol 2024; 86:127516. [PMID: 39226872 DOI: 10.1016/j.jtemb.2024.127516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/26/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Myocardial infarction (MI) is the result of reduced or stopped blood supply to a section of the myocardium. Regardless of its potential effectiveness in the treatment of several types of cancers, doxorubicin (DOX) capabilities are restricted because of its widespread cardiotoxic impact. AIM In this study, the protective effect of zinc oxide nanoparticles against doxorubicin-induced myocardial infarction in rats is examined. METHODS Zinc oxide nanoparticles (ZnO NPs) were synthesized and characterized using X-ray diffraction, transmission electron microscope, and UV-Vis spectral analysis. A total cumulative dose of DOX (18 mg/kg body weight, i.p.) was injected once daily on days 2, 4, 6, 8, 10, and 12 (i.p.) to induce MI in rats. 24 rats were divided into 4 groups; control, MI, and MI treated with two doses of ZnO NPs (45 and 22.5 mg/kg). RESULTS The treatment with ZnO NPs restored ST-segment near normal, ameliorated the changes in cardiac troponin T, creatine kinase, lactate dehydrogenase, aspartate aminotransferase, alanine amino transferase, alkaline phosphatase, total proteins, malondialdehyde, nitric oxide, reduced glutathione, and catalase.The histological investigation revealed that ZnO NPs treated group showed marked improvement in the examined cardiac muscle and liver in numerous sections.The lower dose of ZnO NPs (22.5 mg/kg) was significantly more effective than the higher dose (45 mg/kg). CONCLUSION The effect of ZnO NPs against doxorubicin-induced myocardial infarction in rats was assessed and the results revealed a successful cardioprotective potency through enhancing the antioxidant system and stimulating nitric oxide production in myocardial infarcted rats. This work implies that ZnO NPs could serve as promising agents for treating doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
| | - Mohamed A Ragheb
- Chemistry Department (Biochemistry Division), Faculty of Science, Cairo University, Egypt.
| | | | | |
Collapse
|
14
|
Zhang J, Wang S, Sun Q, Zhang J, Shi X, Yao M, Chen J, Huang Q, Zhang G, Huang Q, Ai K, Bai Y. Peroxynitrite-Free Nitric Oxide-Embedded Nanoparticles Maintain Nitric Oxide Homeostasis for Effective Revascularization of Myocardial Infarcts. ACS NANO 2024; 18:32650-32671. [PMID: 39545833 DOI: 10.1021/acsnano.4c10118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Revascularization is crucial for treating myocardial infarction (MI). Nitric oxide (NO), at an appropriate concentration, is recognized as an ideal and potent pro-angiogenic factor. However, the application of NO in the treatment of MI is limited. Improper NO supplementation is harmful to revascularization because NO is converted into harmful peroxynitrite (ONOO-) in MI tissues with high reactive oxygen species (ROS) levels. We overcome these obstacles by embedding biliverdin and NO into Prussian blue (PB) nanolattices to obtain an ONOO--free NO-embedded nanomedicine (OFEN). Unlike previous NO donors, OFEN provides NO stably and spontaneously for a longer time (>7 days), which makes it possible to maintain a stable concentration of NO, suitable for angiogenesis, through dose optimization. More importantly, based on the synergy between PB and biliverdin, OFEN converts ROS into beneficial O2 and inhibits the production of ONOO- from the source. OFEN specifically targets MI tissues and achieves sustained and stable NO delivery at the MI site. OFEN effectively promotes revascularization in the MI tissue, significantly reduces myocardial death and fibrosis, and ultimately promotes the complete recovery of cardiac function. Our strategy provides a promising approach for the treatment of myocardial and other ischemic diseases.
Collapse
Affiliation(s)
- Jiaxiong Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Quan Sun
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jian Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Meilian Yao
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Jing Chen
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Guogang Zhang
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410000, PR China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, PR China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, PR China
| | - Yongping Bai
- Department of Geriatric Medicine, Coronary Circulation Center, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| |
Collapse
|
15
|
Hu X, Xu H, Bu L, Sun J, Deng J, Song K, Wang L, Pang B. Exploring the wound healing potential of dietary nitrate in diabetic rat model. Front Physiol 2024; 15:1475375. [PMID: 39633648 PMCID: PMC11614883 DOI: 10.3389/fphys.2024.1475375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction The wound healing in diabetes is hindered and prolonged due to long-term inflammation, oxidative stress damage, and angiogenesis disorders induced by high glucose status. The management of such difficult-to-treat wounds continues to pose a significant challenge in clinical treatment. Dietary nitrate, commonly found in greens such as beets and spinach, acts as a nutritional supplement and is metabolized in the body through the salivary nitrate-nitrite-NO pathway. This pathway plays a crucial role in various physiological functions, including enhancing blood flow and attenuating inflammation. Methods In this study, we established a diabetic rat wound model. Forty-eight rats were randomly divided into six groups (n = 8): the Con group, the Con + Nitrate group, the STZ group, the STZ + NaCl group, the STZ + rhEGF group, and the STZ + Nitrate group. Skin wound healing was assessed on the day of surgery and on postoperative days 3, 7, 10, and 14. Specimens were taken on days 7 and 14 post-surgery for relevant tests. Results We found that dietary nitrate could accelerate skin wound healing by promoting angiogenesis and increasing blood perfusion. Significantly, dietary nitrate also regulated glucose and lipid metabolism and exhibited anti-inflammatory and antioxidant properties. Discussion These findings provide a novel theoretical basis for managing wounds in diabetic individuals, indicating the broad potential of dietary nitrate in future clinical applications.
Collapse
Affiliation(s)
- Xiaodan Hu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Haoyue Xu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Lingxue Bu
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Jian Sun
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Jiangzhi Deng
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Kai Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Lin Wang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| | - Baoxing Pang
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Stomatology of Qingdao University, Qingdao, China
| |
Collapse
|
16
|
Yin W, Chen Y, Wang W, Guo M, Tong L, Zhang M, Wang Z, Yuan H. Macrophage-mediated heart repair and remodeling: A promising therapeutic target for post-myocardial infarction heart failure. J Cell Physiol 2024; 239:e31372. [PMID: 39014935 DOI: 10.1002/jcp.31372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Heart failure (HF) remains prevalent in patients who survived myocardial infarction (MI). Despite the accessibility of the primary percutaneous coronary intervention and medications that alleviate ventricular remodeling with functional improvement, there is an urgent need for clinicians and basic scientists to further reveal the mechanisms behind post-MI HF as well as investigate earlier and more efficient treatment after MI. Growing numbers of studies have highlighted the crucial role of macrophages in cardiac repair and remodeling following MI, and timely intervention targeting the immune response via macrophages may represent a promising therapeutic avenue. Recently, technology such as single-cell sequencing has provided us with an updated and in-depth understanding of the role of macrophages in MI. Meanwhile, the development of biomaterials has made it possible for macrophage-targeted therapy. Thus, an overall and thorough understanding of the role of macrophages in post-MI HF and the current development status of macrophage-based therapy will assist in the further study and development of macrophage-targeted treatment for post-infarction cardiac remodeling. This review synthesizes the spatiotemporal dynamics, function, mechanism and signaling of macrophages in the process of HF after MI, as well as discusses the emerging bio-materials and possible therapeutic agents targeting macrophages for post-MI HF.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yong Chen
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lingjun Tong
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaoyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
17
|
Jia X, Liu W, Ai Y, Cheung S, Hu W, Wang Y, Shi X, Zhou J, Zhang Z, Liang Q. A Multifunctional Anisotropic Patch Manufactured by Microfluidic Manipulation for the Repair of Infarcted Myocardium. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404071. [PMID: 39279582 DOI: 10.1002/adma.202404071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/20/2024] [Indexed: 09/18/2024]
Abstract
Engineered hydrogel patches have shown promising therapeutic effects in the treatment of myocardial infarction (MI), especially anisotropic patches that mimic the characteristics of native myocardium have attracted widespread attention. However, it remains a great challenge to develop cardiac patches with long-range and orderly electrical conduction based on an effective, mild, and rapid strategy. Here, a multifunctional anisotropic cardiac patch is presented based on microfluidic manipulation. The anisotropic alginate-gelatin methacrylate hydrogel patches are easily and rapidly prepared through microfluidic focusing, ion-photocrosslinking, and parallel packing processes. The fluid-based anisotropic realization process does not involve complex machining and strong field stimulation and is compatible with the loading of macromolecular biological agents. The anisotropic hydrogel patch can mimic the anisotropy of the myocardium and guide the directional polarization of cardiomyocytes. In animal model experiments, it also exhibits significant effects in inhibiting ventricular remodeling, fibrosis, and enhancing cardiac function recovery after MI. These comprehensive features make the multifunctional hydrogel patch a promising candidate for cardiac tissue repair and future provide a new paradigm for expanding microfluidic technology to solve tissue engineering challenges.
Collapse
Affiliation(s)
- Xiaomeng Jia
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Weiyu Liu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Yongjian Ai
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Suet Cheung
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Yu Wang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| | - Xiaolu Shi
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jin Zhou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Zhuang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Laboratory of Flexible Electronics Technology, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, P. R. China
| |
Collapse
|
18
|
Zhang Y, Li J, Pan J, Deng S. Research progress of two-pore potassium channel in myocardial ischemia-reperfusion injury. Front Physiol 2024; 15:1473501. [PMID: 39534859 PMCID: PMC11554511 DOI: 10.3389/fphys.2024.1473501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a secondary injury caused by restoring blood flow after acute myocardial infarction, which may lead to serious arrhythmia and heart damage. In recent years, the role of potassium channels in MIRI has attracted much attention, especially the members of the two-pore domain potassium (K2P) channel family. K2P channel has unique structure and function, and the formation of its heterodimer increases its functional diversity. This paper reviews the structural characteristics, types, expression and physiological functions of K2P channel in the heart. In particular, we pay attention to whether members of the subfamily such as TWIK, TREK, TASK, TALK, THIK and TRESK participate in MIRI and their related mechanisms. Future research will help to reveal the molecular mechanism of K2P channel in MIRI and provide new strategies for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Shengli Deng
- Department of Anesthesiology, The Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
19
|
Gan G, Shen Z, Zheng S, Zhang G, Yin D, Liu S, Hu J. Biomimetic Activation of N-Nitrosamides with Red Light-Triggered Nitric Oxide Release via Mediated Electron Transfer. Angew Chem Int Ed Engl 2024; 63:e202409981. [PMID: 39037730 DOI: 10.1002/anie.202409981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 07/23/2024]
Abstract
Mediated electron transfer (MET) is fundamental to many biological functions, including cellular respiration, photosynthesis, and enzymatic catalysis. However, leveraging the MET process to enable the release of therapeutic gases has been largely unexplored. Herein, we report the bio-inspired activation of a series of UV-absorbing N-nitrosamide derivatives (NOA) under red light exposure, enabling the quantitative release of nitric oxide (NO) gasotransmitter via an MET process. The cornerstone of our design is the covalent linkage of a 2,4-dinitroaniline moiety, which acts as an electron mediator to the N-nitrosamide groups. This facilitates efficient electron transfer from the excited palladium(II) meso-tetraphenyltetrabenzoporphyrin (PdTPTBP) photocatalyst and the selective activation of NOA. Our approach has been validated with distinct photocatalysts and various N-nitrosamides, including those derived from carbamates, amides, and ureas. Notably, the modulation of the linker length between the electron mediator and N-nitrosamide groups serves as a regulatory mechanism for controlling NO release kinetics. Moreover, this biomimetic NO release platform demonstrates effective operation under both normoxic and hypoxic conditions, and it enables localized delivery of NO under physiological conditions, exhibiting significant anticancer efficacy within the phototherapeutic window and enhanced selectivity towards tumor cells.
Collapse
Affiliation(s)
- Guihai Gan
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| | - Zhiqiang Shen
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| | - Shaoqiu Zheng
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| | - Guoying Zhang
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| | - Dalong Yin
- Department of Hepatobiliary Surgery and Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of University of Science and Technology of China (USTC), and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui Province, 230026, China
| |
Collapse
|
20
|
Cao Y, Fan R, Zhu K, Gao Y. Advances in Functionalized Hydrogels in the Treatment of Myocardial Infarction and Drug-Delivery Strategies. ACS APPLIED MATERIALS & INTERFACES 2024; 16:48880-48894. [PMID: 39227344 DOI: 10.1021/acsami.4c09623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Myocardial infarction (MI) is a serious cardiovascular disease with high morbidity and mortality rates, posing a significant threat to patient's health and quality of life. Following a MI, the damaged myocardial tissue is typically not fully repaired, leading to permanent impairment of myocardial function. While traditional treatments can alleviate symptoms and reduce pain, their ability to repair damaged heart muscle tissue is limited. Functionalized hydrogels, a broad category of materials with diverse functionalities, can enhance the properties of hydrogels to cater to the needs of tissue engineering, drug delivery, medical dressings, and other applications. Recently, functionalized hydrogels have emerged as a promising new therapeutic approach for the treatment of MI. Functionalized hydrogels possess outstanding biocompatibility, customizable mechanical properties, and drug-release capabilities. These properties enable them to offer scaffold support, drug release, and tissue regeneration promotion, making them a promising approach for treating MI. This paper aims to evaluate the advancements and delivery methods of functionalized hydrogels for treating MI, while also discussing their potential and the challenges they may pose for future clinical use.
Collapse
Affiliation(s)
- Yuchen Cao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Rong Fan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Kaiyi Zhu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan 030032, China
| | - Yuping Gao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
- Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan 030032, China
- Key Laboratory of Cellular Physiology, Shanxi Province, Taiyuan 030032, China
| |
Collapse
|
21
|
Niu Q, Li D, Guo W, Feng Z, Han Z, Yang Y. Dietary nitrate maintains homeostasis of oxidative stress and gut microbiota to promote flap survival in type 2 diabetes mellitus rats. BMC Endocr Disord 2024; 24:184. [PMID: 39256735 PMCID: PMC11386097 DOI: 10.1186/s12902-024-01691-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/13/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Random-pattern skin flaps are commonly used to repair skin tissue defects in surgical tissue reconstruction. However, flap necrosis in the distal area due to ischemia injury is still challenging for its applications in plastic surgery. The complications of diabetes will further increase the risk of infection and necrosis. METHODS This study induced type 2 diabetes mellitus (T2DM) rats with a high-fat diet and STZ. The survival rate of the skin flap was observed by adding inorganic sodium nitrate to drinking water. Histology and immunohistochemistry were used to detect the damage to the skin flap. The nitrate content was measured by total nitric oxide and nitrate/nitrite parameter assay. Dihydroethidium and malondialdehyde (MDA) assays were used to value oxidative stress. Rat colon feces were collected for 16s rRNA gene sequence. RESULTS Our studies showed that nitrate administration leads to anti-obesity and anti-diabetic effects. Nitrate directly increased the survival area of skin flaps in diabetic rats and mean blood vessel density by enhancing angiogenesis, inhibiting apoptosis, and reducing oxidative stress. The 16s rRNA sequence revealed that nitrate may regulate the homeostasis of the gut microbiota and re-store energy metabolism. CONCLUSION Dietary nitrate has been shown to maintain the homeostasis of oxidative stress and gut microbiota to promote flap survival in rats with T2DM.
Collapse
Affiliation(s)
- Qifang Niu
- Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Delong Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Wenwen Guo
- Department of Stomatology, First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhien Feng
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Zhengxue Han
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China
| | - Yang Yang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
22
|
Wang Y, Chen X, Chen Z, Wang X, Wang H, Zhai H, Ding J, Yu L. Autophagy inhibition mediated via an injectable and NO-releasing hydrogel for amplifying the antitumor efficacy of mild magnetic hyperthermia. Bioact Mater 2024; 39:336-353. [PMID: 38827171 PMCID: PMC11140189 DOI: 10.1016/j.bioactmat.2024.05.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/05/2024] [Accepted: 05/17/2024] [Indexed: 06/04/2024] Open
Abstract
While mild hyperthermia holds great potential in the treatment of solid tumors, the thermal stress-triggered self-repairing autophagy significantly compromises its efficacy. To circumvent this obstacle, an injectable hydrogel (NO-Gel) composed of thermosensitive poly(ethylene glycol)-polypeptide copolymers modified with abundant NO donors on their side chains is developed. Meanwhile, ferrimagnetic Zn0.5Fe2.5O4 magnetic nanoparticles (MNPs) with high magnetic-heat conversion efficiency are synthesized and loaded into NO-Gel to obtain MNPs@NO-Gel. The MNPs@NO-Gel system exhibits a sol-gel transition upon heating, and has the ability to perform multiple magnetic hyperthermia therapy (MHT) after only one administration due to the even distribution and strong immobilization of MNPs in NO-Gel. NO can be continuously liberated from NO-Gel and this process is markedly accelerated by MHT. Additionally, MNPs@NO-Gel maintains its integrity in vivo for over one month and the released MNPs are metabolized by the spleen. After a single administration of MNPs@NO-Gel at the tumor site, three mild MHT treatments with similar effects are fulfilled, and the sufficient supply of NO effectively inhibits MHT-induced autophagic flux via blocking the formation of autophagosomes and synchronously destroying lysosomes, thereby substantially boosting the efficacy of mild MHT. As a consequence, CT-26 colon tumors are completely eliminated without causing severe side-effects.
Collapse
Affiliation(s)
- Yaoben Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Xiaobin Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Zhiyong Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Xin Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Hancheng Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Huajuan Zhai
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200438, China
| |
Collapse
|
23
|
Han D, Wang F, Shen D. Nanomedicines as Guardians of the Heart: Unleashing the Power of Antioxidants to Alleviate Myocardial Ischemic Injury. Theranostics 2024; 14:5336-5370. [PMID: 39267789 PMCID: PMC11388064 DOI: 10.7150/thno.99961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
Ischemic heart disease (IHD) is increasingly recognized as a significant cardiovascular disease with a growing global incidence. Interventions targeting the oxidative microenvironment have long been pivotal in therapeutic strategies. However, many antioxidant drugs face limitations due to pharmacokinetic and delivery challenges, such as short half-life, poor stability, low bioavailability, and significant side effects. Fortunately, nanotherapies exhibit considerable potential in addressing IHD. Nanomedicines offer advantages such as passive/active targeting, prolonged circulation time, enhanced bioavailability, and diverse carrier options. This comprehensive review explores the advancements in nanomedicines for mitigating IHD through oxidative stress regulation, providing an extensive overview for researchers in the field of antioxidant nanomedicines. By inspiring further research, this study aims to accelerate the development of novel therapies for myocardial injury.
Collapse
Affiliation(s)
- Dongjian Han
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Fuhang Wang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| | - Deliang Shen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Cardiac Injury and Repair of Henan Province, Zhengzhou, China
| |
Collapse
|
24
|
Rayat Pisheh H, Nojabaei FS, Darvishi A, Rayat Pisheh A, Sani M. Cardiac tissue engineering: an emerging approach to the treatment of heart failure. Front Bioeng Biotechnol 2024; 12:1441933. [PMID: 39211011 PMCID: PMC11357970 DOI: 10.3389/fbioe.2024.1441933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Heart failure is a major health problem in which the heart is unable to pump enough blood to meet the body's needs. It is a progressive disease that becomes more severe over time and can be caused by a variety of factors, including heart attack, cardiomyopathy and heart valve disease. There are various methods to cure this disease, which has many complications and risks. The advancement of knowledge and technology has proposed new methods for many diseases. One of the promising new treatments for heart failure is tissue engineering. Tissue engineering is a field of research that aims to create living tissues and organs to replace damaged or diseased tissue. The goal of tissue engineering in heart failure is to improve cardiac function and reduce the need for heart transplantation. This can be done using the three important principles of cells, biomaterials and signals to improve function or replace heart tissue. The techniques for using cells and biomaterials such as electrospinning, hydrogel synthesis, decellularization, etc. are diverse. Treating heart failure through tissue engineering is still under development and research, but it is hoped that there will be no transplants or invasive surgeries in the near future. In this study, based on the most important research in recent years, we will examine the power of tissue engineering in the treatment of heart failure.
Collapse
Affiliation(s)
- Hossein Rayat Pisheh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Sadat Nojabaei
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Science, Tehran, Iran
| | - Ahmad Darvishi
- School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Rayat Pisheh
- Department of Biology, Payam Noor University (PUN), Shiraz, Iran
| | - Mahsa Sani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
25
|
Liu T, Hao Y, Zhang Z, Zhou H, Peng S, Zhang D, Li K, Chen Y, Chen M. Advanced Cardiac Patches for the Treatment of Myocardial Infarction. Circulation 2024; 149:2002-2020. [PMID: 38885303 PMCID: PMC11191561 DOI: 10.1161/circulationaha.123.067097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Myocardial infarction is a cardiovascular disease characterized by a high incidence rate and mortality. It leads to various cardiac pathophysiological changes, including ischemia/reperfusion injury, inflammation, fibrosis, and ventricular remodeling, which ultimately result in heart failure and pose a significant threat to global health. Although clinical reperfusion therapies and conventional pharmacological interventions improve emergency survival rates and short-term prognoses, they are still limited in providing long-lasting improvements in cardiac function or reversing pathological progression. Recently, cardiac patches have gained considerable attention as a promising therapy for myocardial infarction. These patches consist of scaffolds or loaded therapeutic agents that provide mechanical reinforcement, synchronous electrical conduction, and localized delivery within the infarct zone to promote cardiac restoration. This review elucidates the pathophysiological progression from myocardial infarction to heart failure, highlighting therapeutic targets and various cardiac patches. The review considers the primary scaffold materials, including synthetic, natural, and conductive materials, and the prevalent fabrication techniques and optimal properties of the patch, as well as advanced delivery strategies. Last, the current limitations and prospects of cardiac patch research are considered, with the goal of shedding light on innovative products poised for clinical application.
Collapse
Affiliation(s)
- Tailuo Liu
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Ying Hao
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Zixuan Zhang
- West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, PR China (Z.Z.)
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases (T.L., Y.H., H.Z., S.P., D.Z., Y.C., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Shiqin Peng
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Dingyi Zhang
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| | - Ka Li
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Yuwen Chen
- Medicine and Engineering Interdisciplinary Research Laboratory of Nursing & Materials, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, PR China (T.L., K.L., Y.C.)
| | - Mao Chen
- Department of Cardiology (T.L., S.P., D.Z., M.C.), West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
26
|
Chen G, Yu J, Wu L, Ji X, Xu J, Wang C, Ma S, Miao Q, Wang L, Wang C, Lewis SE, Yue Y, Sun Z, Liu Y, Tang B, James TD. Fluorescent small molecule donors. Chem Soc Rev 2024; 53:6345-6398. [PMID: 38742651 PMCID: PMC11181996 DOI: 10.1039/d3cs00124e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Indexed: 05/16/2024]
Abstract
Small molecule donors (SMDs) play subtle roles in the signaling mechanism and disease treatments. While many excellent SMDs have been developed, dosage control, targeted delivery, spatiotemporal feedback, as well as the efficiency evaluation of small molecules are still key challenges. Accordingly, fluorescent small molecule donors (FSMDs) have emerged to meet these challenges. FSMDs enable controllable release and non-invasive real-time monitoring, providing significant advantages for drug development and clinical diagnosis. Integration of FSMDs with chemotherapeutic, photodynamic or photothermal properties can take full advantage of each mode to enhance therapeutic efficacy. Given the remarkable properties and the thriving development of FSMDs, we believe a review is needed to summarize the design, triggering strategies and tracking mechanisms of FSMDs. With this review, we compiled FSMDs for most small molecules (nitric oxide, carbon monoxide, hydrogen sulfide, sulfur dioxide, reactive oxygen species and formaldehyde), and discuss recent progress concerning their molecular design, structural classification, mechanisms of generation, triggered release, structure-activity relationships, and the fluorescence response mechanism. Firstly, from the large number of fluorescent small molecular donors available, we have organized the common structures for producing different types of small molecules, providing a general strategy for the development of FSMDs. Secondly, we have classified FSMDs in terms of the respective donor types and fluorophore structures. Thirdly, we discuss the mechanisms and factors associated with the controlled release of small molecules and the regulation of the fluorescence responses, from which universal guidelines for optical properties and structure rearrangement were established, mainly involving light-controlled, enzyme-activated, reactive oxygen species-triggered, biothiol-triggered, single-electron reduction, click chemistry, and other triggering mechanisms. Fourthly, representative applications of FSMDs for trackable release, and evaluation monitoring, as well as for visible in vivo treatment are outlined, to illustrate the potential of FSMDs in drug screening and precision medicine. Finally, we discuss the opportunities and remaining challenges for the development of FSMDs for practical and clinical applications, which we anticipate will stimulate the attention of researchers in the diverse fields of chemistry, pharmacology, chemical biology and clinical chemistry. With this review, we hope to impart new understanding thereby enabling the rapid development of the next generation of FSMDs.
Collapse
Affiliation(s)
- Guang Chen
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Jing Yu
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Luling Wu
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
| | - Xinrui Ji
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Jie Xu
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Chao Wang
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Siyue Ma
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Qing Miao
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Linlin Wang
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Chen Wang
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Simon E Lewis
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
| | - Yanfeng Yue
- Department of Chemistry, Delaware State University, Dover, DE, 19901, USA.
| | - Zhe Sun
- Institute of Molecular Plus, Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Tianjin University, 92 Weijin Road, Tianjin, 300072, China.
| | - Yuxia Liu
- The Youth Innovation Team of Shaanxi Universities, Shaanxi Key Laboratory of Chemical Additives for Industry, College of Chemistry and Chemical Engineering, Shaanxi University of Science & Technology, Xi'an, 710021, China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan 250014, Shandong, China.
| | - Tony D James
- Department of Chemistry, University of Bath, Bath BA2 7AY, UK.
- School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
27
|
Kenawy ER, Abd El Hay AM, Saad N, Azaam MM, Shoueir KR. Synthesis, characterization of poly L(+) lactic acid and its application in sustained release of isosorbide dinitrate. Sci Rep 2024; 14:7062. [PMID: 38528000 DOI: 10.1038/s41598-024-56222-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 03/04/2024] [Indexed: 03/27/2024] Open
Abstract
Poly L(+) lactic acid (PLLA) has become crucial in the biomedical industry for various uses. The direct polycondensation method was used to prepare Poly L(+) Lactic Acid (PLLA). Different catalysts, including metal oxides and metal halides, were used to test the polymerization technique. The effect of the amount of catalysts and the type of coupling agent were investigated. The effect of reaction time and polymerization solvents was also studied. PLLA was loaded with isosorbide dinitrate utilizing the solvent evaporation process. The synthesized polymer-drug system was evaluated by different means such as FT-IR, TGA, DSC, XRD, entrapment efficiency (E.E), drug loading (D.L), particle size analysis, and zeta potential determination. Studies on in-vitro release using UV light at 227 nm at various pH levels were conducted, and the kinetics of release and cytotoxicity using the sulforhodamine B (SRB) assay on human skin fibroblast cells were examined.
Collapse
Affiliation(s)
- El-Refaie Kenawy
- Polymer Research Group, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - A M Abd El Hay
- Chemistry Department, Faculty of Science, Al Azhar University, Cairo, Egypt
- Regional Technical Manager, Pharma Business Group, IMCD, Cairo, Egypt
| | - Nermeen Saad
- Polymer Research Group, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mohamed M Azaam
- Polymer Research Group, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Kamel R Shoueir
- Institute of Nanoscience and Nanotechnology, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
| |
Collapse
|
28
|
Liu Y, Wang L, Liu Z, Kang Y, Chen T, Xu C, Zhu T. Durable Immunomodulatory Nanofiber Niche for the Functional Remodeling of Cardiovascular Tissue. ACS NANO 2024; 18:951-971. [PMID: 38146717 DOI: 10.1021/acsnano.3c09692] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Functional remodeling and prolonged anti-inflammatory responses are both vital for repairing damage in the cardiovascular system. Although these aspects have each been studied extensively alone, attempts to fabricate scaffolds that combine these effects have seen limited success. In this study, we synthesized salvianic acid A (SA, danshensu) blocked biodegradable polyurethane (PCHU-D) and enclosed it within electrospun nanofibers to synthesize a durable immunomodulatory nanofiber niche (DINN), which provided sustained SA release during inflammation. Given its excellent processability, mechanical properties, and shape memory function, we developed two variants of the DINN as vascular scaffolds and heart patches. Both these variants exhibited outstanding therapeutic effects in in vivo experiments. The DINN was expertly designed such that it gradually decomposes along with SA release, substantially facilitating cellular infiltration and tissue remodeling. Therefore, the DINN effectively inhibited the migration and chemotaxis of inflammatory cells, while also increasing the expression of angiogenic genes. As a result, it promoted the recovery of myocardial function after myocardial infarction and induced rapid reendothelialization following arterial orthotopic transplantation repair. These excellent characteristics indicate that the DINN holds great potential as a multifunctional agent for repairing cardiovascular tissues.
Collapse
Affiliation(s)
- Yonghang Liu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P.R. China
| | - Liren Wang
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, P.R. China
| | - Zhuo Liu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, 138 Xueyuan Road, Shanghai 200032, P.R. China
| | - Yuhao Kang
- Department of Sports Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai 200233, P.R. China
| | - Tianhui Chen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai 200031, P.R. China
| | - Chen Xu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, School of Basic Medical Sciences, Fudan University, 138 Xueyuan Road, Shanghai 200032, P.R. China
| | - Tonghe Zhu
- Multidisciplinary Centre for Advanced Materials, Institute for Frontier Medical Technology, School of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, 333 Longteng Road, Shanghai 201620, P.R. China
| |
Collapse
|
29
|
Zhou Z, Liu Y, Li W, Zhao Z, Xia X, Liu J, Deng Y, Wu Y, Pan X, He F, Yang H, Lu W, Xu Y, Zhu X. A Self-Adaptive Biomimetic Periosteum Employing Nitric Oxide Release for Augmenting Angiogenesis in Bone Defect Regeneration. Adv Healthc Mater 2024; 13:e2302153. [PMID: 37922941 DOI: 10.1002/adhm.202302153] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/12/2023] [Indexed: 11/07/2023]
Abstract
The periosteum plays a vital role in the regeneration of critical-size bone defects and highly comminuted fractures, promoting the differentiation of osteoblasts, accelerating the reconstruction of the vascular network, and guiding bone tissue regeneration. However, the materials loaded with exogenous growth factors are limited by the release and activity of the elements. Therefore, the material structure must be carefully designed for the periosteal function. Here, a self-adaptive biomimetic periosteum strategy is proposed, which is a novel interpenetrating double network hydrogel consisting of diselenide-containing gelatin and calcium alginate (modified natural collagen and polysaccharide) to enhance the stability, anti-swelling, and delayed degradation of the hydrogel. The diselenide bond continuously releases nitric oxide (NO) by metabolizing endogenous nitrosated thiols (RSNO), activates the nitric oxide-cycle guanosine monophosphate (NO-cGMP) signal pathway, coordinates the coupling effect of angiogenesis and osteogenesis, and accelerates the repair of bone defects. This self-adaptive biomimetic periosteum with the interpenetrating double network structure formed by the diselenide-containing gelatin and calcium alginate has been proven to be safe and effective in repairing critical-size bone defects and is expected to provide a promising strategy for solving clinical problems.
Collapse
Affiliation(s)
- Zhangzhe Zhou
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yang Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Wenjing Li
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhijian Zhao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Junlin Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yaoge Deng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Yubin Wu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xiangqiang Pan
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Fan He
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Weihong Lu
- Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Department of Polymer Science and Engineering, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Yong Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| | - Xuesong Zhu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, 215007, China
| |
Collapse
|
30
|
Lee M, Kim YS, Park J, Choe G, Lee S, Kang BG, Jun JH, Shin Y, Kim M, Ahn Y, Lee JY. A paintable and adhesive hydrogel cardiac patch with sustained release of ANGPTL4 for infarcted heart repair. Bioact Mater 2024; 31:395-407. [PMID: 37680586 PMCID: PMC10481188 DOI: 10.1016/j.bioactmat.2023.08.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/05/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
The infarcted heart undergoes irreversible pathological remodeling after reperfusion involving left ventricle dilation and excessive inflammatory reactions in the infarcted heart, frequently leading to fatal functional damage. Extensive attempts have been made to attenuate pathological remodeling in infarcted hearts using cardiac patches and anti-inflammatory drug delivery. In this study, we developed a paintable and adhesive hydrogel patch using dextran-aldehyde (dex-ald) and gelatin, incorporating the anti-inflammatory protein, ANGPTL4, into the hydrogel for sustained release directly to the infarcted heart to alleviate inflammation. We optimized the material composition, including polymer concentration and molecular weight, to achieve a paintable, adhesive hydrogel using 10% gelatin and 5% dex-ald, which displayed in-situ gel formation within 135 s, cardiac tissue-like modulus (40.5 kPa), suitable tissue adhesiveness (4.3 kPa), and excellent mechanical stability. ANGPTL4 was continuously released from the gelatin/dex-ald hydrogel without substantial burst release. The gelatin/dex-ald hydrogel could be conveniently painted onto the beating heart and degraded in vivo. Moreover, in vivo studies using animal models of acute myocardial infarction revealed that our hydrogel cardiac patch containing ANGPTL4 significantly improved heart tissue repair, evaluated by echocardiography and histological evaluation. The heart tissues treated with ANGPTL4-loaded hydrogel patches exhibited increased vascularization, reduced inflammatory macrophages, and structural maturation of cardiac cells. Our novel hydrogel system, which allows for facile paintability, appropriate tissue adhesiveness, and sustained release of anti-inflammatory drugs, will serve as an effective platform for the repair of various tissues, including heart, muscle, and cartilage.
Collapse
Affiliation(s)
- Mingyu Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Yong Sook Kim
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Sanghun Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Bo Gyeong Kang
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
| | - Ju Hee Jun
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
| | - Yoonmin Shin
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Minchul Kim
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youngkeun Ahn
- Cell Regeneration Research Center, Chonnam National University, Gwangju, Republic of Korea
- Department of Cardiology, Chonnam National University Hospital, Gwangju, Republic of Korea
- Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| |
Collapse
|
31
|
Liu X, Chen B, Chen J, Wang X, Dai X, Li Y, Zhou H, Wu LM, Liu Z, Yang Y. A Cardiac-Targeted Nanozyme Interrupts the Inflammation-Free Radical Cycle in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308477. [PMID: 37985164 DOI: 10.1002/adma.202308477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/14/2023] [Indexed: 11/22/2023]
Abstract
Severe systemic inflammation following myocardial infarction (MI) is a major cause of patient mortality. MI-induced inflammation can trigger the production of free radicals, which in turn ultimately leads to increased inflammation in cardiac lesions (i.e., inflammation-free radicals cycle), resulting in heart failure and patient death. However, currently available anti-inflammatory drugs have limited efficacy due to their weak anti-inflammatory effect and poor accumulation at the cardiac site. Herein, a novel Fe-Cur@TA nanozyme is developed for targeted therapy of MI, which is generated by coordinating Fe3+ and anti-inflammatory drug curcumin (Cur) with further modification of tannic acid (TA). Such Fe-Cur@TA nanozyme exhibits excellent free radicals scavenging and anti-inflammatory properties by reducing immune cell infiltration, promoting macrophage polarization toward the M2-like phenotype, suppressing inflammatory cytokine secretion, and blocking the inflammatory free radicals cycle. Furthermore, due to the high affinity of TA for cardiac tissue, Fe-Cur@TA shows an almost tenfold greater in cardiac retention and uptake than Fe-Cur. In mouse and preclinical beagle dog MI models, Fe-Cur@TA nanozyme preserves cardiac function and reduces scar size, suggesting promising potential for clinical translation in cardiovascular disease.
Collapse
Affiliation(s)
- Xueliang Liu
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Binghua Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jingqi Chen
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xuan Wang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xinfeng Dai
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuqing Li
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Huayuan Zhou
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lian-Ming Wu
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu, 215123, China
| | - Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
32
|
Hao T, Ji G, Qian M, Li QX, Huang H, Deng S, Liu P, Deng W, Wei Y, He J, Wang S, Gao W, Li T, Cheng J, Tian J, Pan L, Gao F, Li Z, Zhao Q. Intracellular delivery of nitric oxide enhances the therapeutic efficacy of mesenchymal stem cells for myocardial infarction. SCIENCE ADVANCES 2023; 9:eadi9967. [PMID: 38019911 PMCID: PMC10686553 DOI: 10.1126/sciadv.adi9967] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Cell therapy by autologous mesenchymal stem cells (MSCs) is a clinically acceptable strategy for treating various diseases. Unfortunately, the therapeutic efficacy is largely affected by the low quality of MSCs collected from patients. Here, we showed that the gene expression of MSCs from patients with diabetes was differentially regulated compared to that of MSCs from healthy controls. Then, MSCs were genetically engineered to catalyze an NO prodrug to release NO intracellularly. Compared to extracellular NO conversion, intracellular NO delivery effectively prolonged survival and enhanced the paracrine function of MSCs, as demonstrated by in vitro and in vivo assays. The enhanced therapeutic efficacy of engineered MSCs combined with intracellular NO delivery was further confirmed in mouse and rat models of myocardial infarction, and a clinically relevant cell administration paradigm through secondary thoracotomy has been attempted.
Collapse
Affiliation(s)
- Tian Hao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Guangbo Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qiu Xuan Li
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haoyan Huang
- Nankai University School of Medicine, Tianjin 300071, China
| | - Shiyu Deng
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Pei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Weiliang Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ju He
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Shusen Wang
- Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, Nankai University, Tianjin, China
| | - Tong Li
- Department of Heart Center, The Third Central Hospital of Tianjin, Nankai University, Tianjin, China
| | - Jiansong Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin 300353, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fei Gao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zongjin Li
- Nankai University School of Medicine, Tianjin 300071, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
33
|
Qian B, Shen A, Huang S, Shi H, Long Q, Zhong Y, Qi Z, He X, Zhang Y, Hai W, Wang X, Cui Y, Chen Z, Xuan H, Zhao Q, You Z, Ye X. An Intrinsically Magnetic Epicardial Patch for Rapid Vascular Reconstruction and Drug Delivery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303033. [PMID: 37964406 PMCID: PMC10754083 DOI: 10.1002/advs.202303033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/30/2023] [Indexed: 11/16/2023]
Abstract
Myocardial infarction (MI) is a major cause of mortality worldwide. The major limitation of regenerative therapy for MI is poor cardiac retention of therapeutics, which results from an inefficient vascular network and poor targeting ability. In this study, a two-layer intrinsically magnetic epicardial patch (MagPatch) prepared by 3D printing with biocompatible materials like poly (glycerol sebacate) (PGS) is designed, poly (ε-caprolactone) (PCL), and NdFeB. The two-layer structure ensured that the MagPatch multifariously utilized the magnetic force for rapid vascular reconstruction and targeted drug delivery. MagPatch accumulates superparamagnetic iron oxide (SPION)-labelled endothelial cells, instantly forming a ready-implanted organization, and rapidly reconstructs a vascular network anastomosed with the host. In addition, the prefabricated vascular network within the MagPatch allowed for the efficient accumulation of SPION-labelled therapeutics, amplifying the therapeutic effects of cardiac repair. This study defined an extendable therapeutic platform for vascularization-based targeted drug delivery that is expected to assist in the progress of regenerative therapies in clinical applications.
Collapse
Affiliation(s)
- Bei Qian
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Ao Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Shixing Huang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Hongpeng Shi
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Qiang Long
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yiming Zhong
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Zhaoxi Qi
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Xiaojun He
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yecen Zhang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Wangxi Hai
- Department of Nuclear Medicine, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Xinming Wang
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Yanna Cui
- Department of Pharmacology and Chemical BiologyShanghai Jiaotong University School of MedicineShanghai200000China
| | - Ziheng Chen
- School of Mechatronics Engineering and AutomationShanghai UniversityShanghai200000China
| | - Huixia Xuan
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Qiang Zhao
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsCollege of Materials Science and EngineeringInstitute of Functional MaterialsResearch Base of Textile Materials for Flexible Electronics and Biomedical Applications (China Textile Engineering Society)Shanghai Engineering Research Center of Nano‐Biomaterials and Regenerative MedicineDonghua UniversityShanghai201620China
| | - Xiaofeng Ye
- Department of Cardiovascular Surgery, Ruijin HospitalShanghai Jiaotong University School of MedicineShanghai200025China
| |
Collapse
|
34
|
Yu Y, Wang S, Chen X, Gao Z, Dai K, Wang J, Liu C. Sulfated oligosaccharide activates endothelial Notch for inducing macrophage-associated arteriogenesis to treat ischemic diseases. Proc Natl Acad Sci U S A 2023; 120:e2307480120. [PMID: 37943835 PMCID: PMC10655224 DOI: 10.1073/pnas.2307480120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 10/01/2023] [Indexed: 11/12/2023] Open
Abstract
Ischemic diseases lead to considerable morbidity and mortality, yet conventional clinical treatment strategies for therapeutic angiogenesis fall short of being impactful. Despite the potential of biomaterials to deliver pro-angiogenic molecules at the infarct site to induce angiogenesis, their efficacy has been impeded by aberrant vascular activation and off-target circulation. Here, we present a semisynthetic low-molecular sulfated chitosan oligosaccharide (SCOS) that efficiently induces therapeutic arteriogenesis with a spontaneous generation of collateral circulation and blood reperfusion in rodent models of hind limb ischemia and myocardial infarction. SCOS elicits anti-inflammatory macrophages' (Mφs') differentiation into perivascular Mφs, which in turn directs artery formation via a cell-to-cell communication rather than secretory factor regulation. SCOS-mediated arteriogenesis requires a canonical Notch signaling pathway in Mφs via the glycosylation of protein O-glucosyltransferases 2, which results in promoting arterial differentiation and tissue repair in ischemia. Thus, this highly bioactive oligosaccharide can be harnessed to direct efficiently therapeutic arteriogenesis and perfusion for the treatment of ischemic diseases.
Collapse
Affiliation(s)
- Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Shuang Wang
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Xinye Chen
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Zehua Gao
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Kai Dai
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| | - Changsheng Liu
- Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai200237, People’s Republic of China
- Key Laboratory for Ultrafine Materials of Ministry of Education, East China University of Science and Technology, Shanghai200237, People’s Republic of China
| |
Collapse
|
35
|
Shen S, Zhang J, Han Y, Pu C, Duan Q, Huang J, Yan B, You X, Lin R, Shen X, Qiu X, Hou H. A Core-Shell Nanoreinforced Ion-Conductive Implantable Hydrogel Bioelectronic Patch with High Sensitivity and Bioactivity for Real-Time Synchronous Heart Monitoring and Repairing. Adv Healthc Mater 2023; 12:e2301990. [PMID: 37467758 DOI: 10.1002/adhm.202301990] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
To achieve synchronous repair and real-time monitoring the infarcted myocardium based on an integrated ion-conductive hydrogel patch is challenging yet intriguing. Herein, a novel synthetic strategy is reported based on core-shell-structured curcumin-nanocomposite-reinforced ion-conductive hydrogel for synchronous heart electrophysiological signal monitoring and infarcted heart repair. The nanoreinforcement and multisite cross-linking of bioactive curcumin nanoparticles enable well elasticity with negligible hysteresis, implantability, ultrahigh mechanoelectrical sensitivity (37 ms), and reliable sensing capacity (over 3000 cycles) for the nanoreinforced hydrogel. Results of in vitro and in vivo experiments demonstrate that such solely physical microenvironment of electrophysiological and biomechanical characteristics combining with the role of bioactive curcumin exert the synchronous benefit of regulating inflammatory microenvironment, promoting angiogenesis, and reducing myocardial fibrosis for effective myocardial infarction (MI) repair. Especially, the hydrogel sensors offer the access for achieving accurate acquisition of cardiac signals, thus monitoring the whole MI healing process. This novel bioactive and electrophysiological-sensing ion-conductive hydrogel cardiac patch highlights a versatile strategy promising for synchronous integration of in vivo real-time monitoring the MI status and excellent MI repair performance.
Collapse
Affiliation(s)
- Si Shen
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jie Zhang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yanni Han
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Chunyi Pu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Qixiang Duan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jianxing Huang
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Bing Yan
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xintong You
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Rurong Lin
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xiaoxi Shen
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Xiaozhong Qiu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Science, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
36
|
Chen C, Zhou Q, Li X, Zhao B, Chen Y, Xiong X. Organic Nitrate Additive for High-Rate and Large-Capacity Lithium Metal Anode in Carbonate Electrolyte. SMALL METHODS 2023:e2300839. [PMID: 37840426 DOI: 10.1002/smtd.202300839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/19/2023] [Indexed: 10/17/2023]
Abstract
Lithium nitrate has been widely used to improve the interfacial stability of Li metal anode in ether electrolyte. However, the low solubility limits its application in carbonate electrolytes for high-voltage Li metal batteries. Herein, nitrated polycaprolactone (PCL-ONO2 ), which is prepared via the acylation of polycaprolactone diol (PCL-diol) followed by the grafting of nitrate group, has been proposed as an electrolyte additive to introduce high-concentration NO3 - into carbonate electrolytes for the first time. The theoretical calculations and X-ray photoelectron spectroscopy depth profiling demonstrate that the PCL-ONO2 additive preferentially reacts with Li metal and in situ constructs a stable dual-layered solid electrolyte interphase film, presenting an inner nitride-rich layer and an outer flexible PCL-based layer on the surface of Li metal anode. As a result, the Li metal anode delivers an impressive long-term cycling performance over 1400 h at an elevated area capacity of 10.0 mAh cm-2 and an ultrahigh current density of 10.0 mA cm-2 in the Li symmetrical cells. Moreover, the PCL-ONO2 additive enables the full cells constructed by coupling high-loading LiFePO4 (20.0 mg cm-2 ) or LiNi0.5 Co0.2 Mn0.3 (16.5 mg cm-2 ) cathode and thin Li metal anode (≈50 µm) to demonstrate greatly improved cycling stability and rate capability.
Collapse
Affiliation(s)
- Chao Chen
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Advanced Energy Storage Materials, South China University of Technology, Guangzhou, 510006, China
| | - Qingfeng Zhou
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Advanced Energy Storage Materials, South China University of Technology, Guangzhou, 510006, China
| | - Xiaodan Li
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Advanced Energy Storage Materials, South China University of Technology, Guangzhou, 510006, China
| | - Bote Zhao
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Advanced Energy Storage Materials, South China University of Technology, Guangzhou, 510006, China
| | - Yunhua Chen
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Xunhui Xiong
- School of Environment and Energy, Guangdong Provincial Key Laboratory of Advanced Energy Storage Materials, South China University of Technology, Guangzhou, 510006, China
| |
Collapse
|
37
|
Andrabi SM, Sharma NS, Karan A, Shahriar SMS, Cordon B, Ma B, Xie J. Nitric Oxide: Physiological Functions, Delivery, and Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303259. [PMID: 37632708 PMCID: PMC10602574 DOI: 10.1002/advs.202303259] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Indexed: 08/28/2023]
Abstract
Nitric oxide (NO) is a gaseous molecule that has a central role in signaling pathways involved in numerous physiological processes (e.g., vasodilation, neurotransmission, inflammation, apoptosis, and tumor growth). Due to its gaseous form, NO has a short half-life, and its physiology role is concentration dependent, often restricting its function to a target site. Providing NO from an external source is beneficial in promoting cellular functions and treatment of different pathological conditions. Hence, the multifaceted role of NO in physiology and pathology has garnered massive interest in developing strategies to deliver exogenous NO for the treatment of various regenerative and biomedical complexities. NO-releasing platforms or donors capable of delivering NO in a controlled and sustained manner to target tissues or organs have advanced in the past few decades. This review article discusses in detail the generation of NO via the enzymatic functions of NO synthase as well as from NO donors and the multiple biological and pathological processes that NO modulates. The methods for incorporating of NO donors into diverse biomaterials including physical, chemical, or supramolecular techniques are summarized. Then, these NO-releasing platforms are highlighted in terms of advancing treatment strategies for various medical problems.
Collapse
Affiliation(s)
- Syed Muntazir Andrabi
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Navatha Shree Sharma
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Anik Karan
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - S. M. Shatil Shahriar
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Brent Cordon
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bing Ma
- Cell Therapy Manufacturing FacilityMedStar Georgetown University HospitalWashington, DC2007USA
| | - Jingwei Xie
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical and Materials EngineeringCollege of EngineeringUniversity of Nebraska LincolnLincolnNE68588USA
| |
Collapse
|
38
|
Wei X, Wang L, Duan C, Chen K, Li X, Guo X, Chen P, Liu H, Fan Y. Cardiac patches made of brown adipose-derived stem cell sheets and conductive electrospun nanofibers restore infarcted heart for ischemic myocardial infarction. Bioact Mater 2023; 27:271-287. [PMID: 37122901 PMCID: PMC10130885 DOI: 10.1016/j.bioactmat.2023.03.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 05/02/2023] Open
Abstract
Cell sheet engineering has been proven to be a promising strategy for cardiac remodeling post-myocardial infarction. However, insufficient mechanical strength and low cell retention lead to limited therapeutic efficiency. The thickness and area of artificial cardiac patches also affect their therapeutic efficiency. Cardiac patches prepared by combining cell sheets with electrospun nanofibers, which can be transplanted and sutured to the surface of the infarcted heart, promise to solve this problem. Here, we fabricated a novel cardiac patch by stacking brown adipose-derived stem cells (BADSCs) sheet layer by layer, and then they were combined with multi-walled carbon nanotubes (CNTs)-containing electrospun polycaprolactone/silk fibroin nanofibers (CPSN). The results demonstrated that BADSCs tended to generate myocardium-like structures seeded on CPSN. Compared with BADSCs suspension-containing electrospun nanofibers, the transplantation of the CPSN-BADSCs sheets (CNBS) cardiac patches exhibited accelerated angiogenesis and decreased inflammation in a rat myocardial infarction model. In addition, the CNBS cardiac patches could regulate macrophage polarization and promote gap junction remodeling, thus restoring cardiac functions. Overall, the hybrid cardiac patches made of electrospun nanofibers and cell sheets provide a novel solution to cardiac remodeling after ischemic myocardial infarction.
Collapse
Affiliation(s)
- Xinbo Wei
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Li Wang
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Cuimi Duan
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Kai Chen
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Xia Li
- Beijing Citident Stomatology Hospital, Beijing, 100032, PR China
| | - Ximin Guo
- Department of Advanced Interdisciplinary Studies, Institute of Basic Medical Sciences, Beijing, 100850, PR China
| | - Peng Chen
- Department of Ultrasound, The Third Medical Center, Chinese PLA General Hospital, Beijing, PR China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, PR China
| |
Collapse
|
39
|
Cao J, Yuan P, Wu B, Liu Y, Hu C. Advances in the Research and Application of Smart-Responsive Hydrogels in Disease Treatment. Gels 2023; 9:662. [PMID: 37623116 PMCID: PMC10454421 DOI: 10.3390/gels9080662] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Smart-responsive hydrogels have been widely used in various fields, particularly in the biomedical field. Compared with traditional hydrogels, smart-responsive hydrogels not only facilitate the encapsulation and controlled release of drugs, active substances, and even cells but, more importantly, they enable the on-demand and controllable release of drugs and active substances at the disease site, significantly enhancing the efficacy of disease treatment. With the rapid advancement of biomaterials, smart-responsive hydrogels have received widespread attention, and a wide variety of smart-responsive hydrogels have been developed for the treatment of different diseases, thus presenting tremendous research prospects. This review summarizes the latest advancements in various smart-responsive hydrogels used for disease treatment. Additionally, some of the current shortcomings of smart-responsive hydrogels and the strategies to address them are discussed, as well as the future development directions and prospects of smart-responsive hydrogels.
Collapse
Affiliation(s)
- Juan Cao
- School of Fashion and Design Art, Sichuan Normal University, Chengdu 610066, China;
| | - Ping Yuan
- School of Mechanical Engineering, Chengdu University, Chengdu 610106, China;
| | - Bo Wu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Yeqi Liu
- School of Mechanical Engineering, Sichuan University, Chengdu 610065, China; (B.W.); (Y.L.)
| | - Cheng Hu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
40
|
Li X, Ou W, Xie M, Yang J, Li Q, Li T. Nanomedicine-Based Therapeutics for Myocardial Ischemic/Reperfusion Injury. Adv Healthc Mater 2023; 12:e2300161. [PMID: 36971662 PMCID: PMC11468948 DOI: 10.1002/adhm.202300161] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/05/2023] [Indexed: 03/29/2023]
Abstract
Myocardial ischemic/reperfusion (IR) injury is a global cardiovascular disease with high mortality and morbidity. Therapeutic interventions for myocardial ischemia involve restoring the occluded coronary artery. However, reactive oxygen species (ROS) inevitably impair the cardiomyocytes during the ischemic and reperfusion phases. Antioxidant therapy holds great promise against myocardial IR injury. The current therapeutic methodologies for ROS scavenging depend predominantly on administering antioxidants. Nevertheless, the intrinsic drawbacks of antioxidants limit their further clinical transformation. The use of nanoplatforms with versatile characteristics greatly benefits drug delivery in myocardial ischemic therapy. Nanoplatform-mediated drug delivery significantly improves drug bioavailability, increases therapeutic index, and reduces systemic toxicity. Nanoplatforms can be specifically and reasonably designed to enhance molecule accumulation at the myocardial site. The present review initially summarizes the mechanism of ROS generation during the process of myocardial ischemia. The understanding of this phenomenon will facilitate the advancement of innovative therapeutic strategies against myocardial IR injury. The latest developments in nanomedicine for treating myocardial ischemic injury are then discussed. Finally, the current challenges and perspectives in antioxidant therapy for myocardial IR injury are addressed.
Collapse
Affiliation(s)
- Xi Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Wei Ou
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
- Department of AnesthesiologyNanchong Central HospitalNanchong637000P. R. China
| | - Maodi Xie
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Jing Yang
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Qian Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| | - Tao Li
- Department of AnesthesiologyLaboratory of Mitochondria and MetabolismNational Clinical Research Center for GeriatricsWest China Hospital of Sichuan UniversityChengdu610041P. R. China
| |
Collapse
|
41
|
Zhao J, Fu J, Jia F, Li J, Yu B, Huang Y, Ren K, Ji J, Fu G. Precise Regulation of Inflammation and Oxidative Stress by ROS‐Responsive Prodrug Coated Balloon for Preventing Vascular Restenosis. ADVANCED FUNCTIONAL MATERIALS 2023; 33. [DOI: 10.1002/adfm.202213993] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Indexed: 09/09/2024]
Abstract
AbstractVascular restenosis after balloon dilation is largely caused by the over‐proliferation of smooth muscle cells, which is triggered and exacerbated by local excessive inflammation and oxidative stress. The excessive inflammatory and oxidative stress cause tissue/cell damage, hamper endothelial functions, and worsen intimal hyperplasia and restenosis. A high level of reactive oxygen species (ROS) overproduction is regarded as the main culprit. Therefore, efficiently inhibiting ROS over‐production or weightily depleting them is of great significance. Herein, a “ROS‐responsive/scavenging prodrug” is introduced into balloon coating for the treatment of vascular restenosis. A reversible phenylboronic ester‐bearing caffeic acid (CA) macromolecular prodrug (PBC) is designed for the controlled and on‐demand dual‐drug release triggered by the local high ROS level; the released CA and 4‐hydroxybenzyl alcohol exhibit efficient antioxidant and anti‐inflammatory effects by scavenging ROS, thereby regulating vascular microenvironment and protecting endothelium functions. To accelerate endothelium regeneration, pro‐endothelial microRNA‐126 is further introduced. The ROS‐responsive/scavenging prodrug/miRNA balloon coating efficiently prevents intimal hyperplasia, alleviates local inflammation, and improves endothelium healing in a rat abdominal aorta restenosis model, which may provide applicative perspectives for next‐generation drug‐coated balloons and other cardiovascular diseases treatment.
Collapse
Affiliation(s)
- Jing Zhao
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
| | - Jia‐yin Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
| | - Fan Jia
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
| | - Jian Li
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
| | - Bo Yu
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Ke‐feng Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Jian Ji
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization Department of Polymer Science and Engineering Zhejiang University Hangzhou 310027 China
| | - Guo‐sheng Fu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province Department of Cardiology Sir Run Run Shaw Hospital Zhejiang University Hangzhou 310016 China
| |
Collapse
|
42
|
Wei Y, Jiang H, Chai C, Liu P, Qian M, Sun N, Gao M, Zu H, Yu Y, Ji G, Zhang Y, Yang S, He J, Cheng J, Tian J, Zhao Q. Endothelium-Mimetic Surface Modification Improves Antithrombogenicity and Enhances Patency of Vascular Grafts in Rats and Pigs. JACC Basic Transl Sci 2023; 8:843-861. [PMID: 37547067 PMCID: PMC10401295 DOI: 10.1016/j.jacbts.2022.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 08/08/2023]
Abstract
We first identified thrombomodulin (TM) and endothelial nitric oxide (NO) synthase as key factors for the antithrombogenic function of the endothelium in human atherosclerotic carotid arteries. Then, recombinant TM and an engineered galactosidase responsible for the conversion of an exogenous NO prodrug were immobilized on the surface of the vascular grafts. Surface modification by TM and NO cooperatively enhanced the antithrombogenicity and patency of vascular grafts. Importantly, we found that the combination of TM and NO also promoted endothelialization, whereas it reduced adverse intimal hyperplasia, which is critical for the maintenance of vascular homeostasis, as confirmed in rat and pig models.
Collapse
Affiliation(s)
- Yongzhen Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Huan Jiang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Chao Chai
- Department of Radiology, Tianjin Institute of Imaging Medicine, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Pei Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Meng Qian
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Na Sun
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia (Ministry of Education), Harbin, China
| | - Man Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Honglin Zu
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Yongquan Yu
- Department of Radiology, Weihai Central Hospital, Weihai, China
| | - Guangbo Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Yating Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| | - Sen Yang
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Ju He
- Department of Vascular Surgery, Tianjin First Central Hospital, Nankai University, Tianjin, China
| | - Jiansong Cheng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Jinwei Tian
- Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia (Ministry of Education), Harbin, China
| | - Qiang Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials (Ministry of Education), Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
43
|
Wang H, Guo Y, Hu Y, Zhou Y, Chen Y, Huang X, Chen J, Deng Q, Cao S, Hu B, Jiang R, Pan J, Tan T, Wang Y, Chen Y, Dong Q, Chen P, Zhou Q. Ultrasound-controlled nano oxygen carriers enhancing cell viability in 3D GelMA hydrogel for the treatment of myocardial infarction. Int J Biol Macromol 2023:125139. [PMID: 37268076 DOI: 10.1016/j.ijbiomac.2023.125139] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/04/2023]
Abstract
Heart failure is a critical and ultimate phase of cardiovascular ailment that leads to a considerable incidence of disability and mortality. Among various factors contributing to heart failure, myocardial infarction is one of the most frequent and significant causes, which is still difficult to manage effectively. An innovative therapeutic strategy, namely a 3D bio-printed cardiac patch, has recently emerged as a promising approach to substitute damaged cardiomyocytes in a localized infarct region. Nevertheless, the efficacy of this treatment primarily relies on the long-term viability of the transplanted cells. In this study, we aimed to construct acoustically sensitive nano oxygen carriers to improve cell survival inside the bio-3D printed patch. In this study, we initially created nanodroplets capable of phase transition triggered by ultrasound and integrated them into GelMA (Gelatin Methacryloyl) hydrogels, which were then employed for 3D bioprinting. After adding nanodroplets and ultrasonic irradiation, numerous pores appeared inside the hydrogel with improved permeability. We further encapsulated hemoglobin into nanodroplets (ND-Hb) to construct oxygen carriers. Results of in vitro experiments showed the highest cell survival within the patch of ND-Hb irradiated by the low-intensity pulsed ultrasound (LIPUS) group. The genomic analysis discovered that the increased survival of seeded cells within the patch might be related to the protection of mitochondrial function owing to the improved hypoxic state. Eventually, in vivo studies revealed that the LIPUS+ND-Hb group had improved cardiac function and increased revascularization after myocardial infarction. To summarize, our study successfully improved the permeability of the hydrogel in a non-invasive and efficient manner, facilitating the exchange of substances in the cardiac patch. Moreover, ultrasound-controlled oxygen release augmented the viability of the transplanted cells and expedited the repair of infarcted tissues.
Collapse
Affiliation(s)
- Hao Wang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yuxin Guo
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yugang Hu
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yanxiang Zhou
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yueying Chen
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xin Huang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jinling Chen
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Qing Deng
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Sheng Cao
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Bo Hu
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Riyue Jiang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Juhong Pan
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Tuantuan Tan
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yijia Wang
- Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Yun Chen
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Qi Dong
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Pu Chen
- Wuhan University School of Basic Medical Science, 430060 Wuhan, China
| | - Qing Zhou
- Renmin Hospital of Wuhan University, 430060 Wuhan, China.
| |
Collapse
|
44
|
Wang Z, Jin A, Yang Z, Huang W. Advanced Nitric Oxide Generating Nanomedicine for Therapeutic Applications. ACS NANO 2023; 17:8935-8965. [PMID: 37126728 PMCID: PMC10395262 DOI: 10.1021/acsnano.3c02303] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Nitric oxide (NO), a gaseous transmitter extensively present in the human body, regulates vascular relaxation, immune response, inflammation, neurotransmission, and other crucial functions. Nitrite donors have been used clinically to treat angina, heart failure, pulmonary hypertension, and erectile dysfunction. Based on NO's vast biological functions, it further can treat tumors, bacteria/biofilms and other infections, wound healing, eye diseases, and osteoporosis. However, delivering NO is challenging due to uncontrolled blood circulation release and a half-life of under five seconds. With advanced biotechnology and the development of nanomedicine, NO donors packaged with multifunctional nanocarriers by physically embedding or chemically conjugating have been reported to show improved therapeutic efficacy and reduced side effects. Herein, we review and discuss recent applications of NO nanomedicines, their therapeutic mechanisms, and the challenges of NO nanomedicines for future scientific studies and clinical applications. As NO enables the inhibition of the replication of DNA and RNA in infectious microbes, including COVID-19 coronaviruses and malaria parasites, we highlight the potential of NO nanomedicines for antipandemic efforts. This review aims to provide deep insights and practical hints into design strategies and applications of NO nanomedicines.
Collapse
Affiliation(s)
- Zhixiong Wang
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Zhen Yang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| | - Wei Huang
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Normal University, Fuzhou, Fujian 350117, China
- Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| |
Collapse
|
45
|
Zhu D, Liu S, Huang K, Li J, Mei X, Li Z, Cheng K. Intrapericardial long non-coding RNA-Tcf21 antisense RNA inducing demethylation administration promotes cardiac repair. Eur Heart J 2023; 44:1748-1760. [PMID: 36916305 PMCID: PMC10411945 DOI: 10.1093/eurheartj/ehad114] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 02/01/2023] [Accepted: 02/16/2023] [Indexed: 03/16/2023] Open
Abstract
AIMS Epicardium and epicardium-derived cells are critical players in myocardial fibrosis. Mesenchymal stem cell-derived extracellular vesicles (EVs) have been studied for cardiac repair to improve cardiac remodelling, but the actual mechanisms remain elusive. The aim of this study is to investigate the mechanisms of EV therapy for improving cardiac remodelling and develop a promising treatment addressing myocardial fibrosis. METHODS AND RESULTS Extracellular vesicles were intrapericardially injected for mice myocardial infarction treatment. RNA-seq, in vitro gain- and loss-of-function experiments, and in vivo studies were performed to identify targets that can be used for myocardial fibrosis treatment. Afterward, a lipid nanoparticle-based long non-coding RNA (lncRNA) therapy was prepared for mouse and porcine models of myocardial infarction treatment. Intrapericardial injection of EVs improved adverse myocardial remodelling in mouse models of myocardial infarction. Mechanistically, Tcf21 was identified as a potential target to improve cardiac remodelling. Loss of Tcf21 function in epicardium-derived cells caused increased myofibroblast differentiation, whereas forced Tcf21 overexpression suppressed transforming growth factor-β signalling and myofibroblast differentiation. LncRNA-Tcf21 antisense RNA inducing demethylation (TARID) that enriched in EVs was identified to up-regulate Tcf21 expression. Formulated lncRNA-TARID-laden lipid nanoparticles up-regulated Tcf21 expression in epicardium-derived cells and improved cardiac function and histology in mouse and porcine models of myocardial infarction. CONCLUSION This study identified Tcf21 as a critical target for improving cardiac fibrosis. Up-regulating Tcf21 by using lncRNA-TARID-laden lipid nanoparticles could be a promising way to treat myocardial fibrosis. This study established novel mechanisms underlying EV therapy for improving adverse remodelling and proposed a lncRNA therapy for cardiac fibrosis.
Collapse
Affiliation(s)
- Dashuai Zhu
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Shuo Liu
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Ke Huang
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Junlang Li
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Xuan Mei
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Zhenhua Li
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| | - Ke Cheng
- Joint Department of Biomedical Engineering, North Carolina State University & University of North Carolina at Chapel Hill, 1001 William Moore Drive, Raleigh, NC 27607, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, 1001 William Moore Drive, Raleigh, NC 27607, USA
| |
Collapse
|
46
|
Gil-Cabrerizo P, Scaccheti I, Garbayo E, Blanco-Prieto MJ. Cardiac tissue engineering for myocardial infarction treatment. Eur J Pharm Sci 2023; 185:106439. [PMID: 37003408 DOI: 10.1016/j.ejps.2023.106439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/26/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Myocardial infarction is one of the major causes of morbidity and mortality worldwide. Current treatments can relieve the symptoms of myocardial ischemia but cannot repair the necrotic myocardial tissue. Novel therapeutic strategies based on cellular therapy, extracellular vesicles, non-coding RNAs and growth factors have been designed to restore cardiac function while inducing cardiomyocyte cycle re-entry, ensuring angiogenesis and cardioprotection, and preventing ventricular remodeling. However, they face low stability, cell engraftment issues or enzymatic degradation in vivo, and it is thus essential to combine them with biomaterial-based delivery systems. Microcarriers, nanocarriers, cardiac patches and injectable hydrogels have yielded promising results in preclinical studies, some of which are currently being tested in clinical trials. In this review, we cover the recent advances made in cellular and acellular therapies used for cardiac repair after MI. We present current trends in cardiac tissue engineering related to the use of microcarriers, nanocarriers, cardiac patches and injectable hydrogels as biomaterial-based delivery systems for biologics. Finally, we discuss some of the most crucial aspects that should be addressed in order to advance towards the clinical translation of cardiac tissue engineering approaches.
Collapse
Affiliation(s)
- Paula Gil-Cabrerizo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain
| | - Ilaria Scaccheti
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, Faculty of Pharmacy and Nutrition, University of Navarra, Pamplona, C/Irunlarrea 1, E-31080, Spain.; Navarra Institute for Health Research, IdiSNA, Pamplona, C/Irunlarrea 3, E-31008 Pamplona, Spain..
| |
Collapse
|
47
|
Das RP, Singh BG, Aishwarya J, Kumbhare LB, Kunwar A. 3,3'-Diselenodipropionic acid immobilised gelatin gel: a biomimic catalytic nitric oxide generating material for topical wound healing application. Biomater Sci 2023; 11:1437-1450. [PMID: 36602012 DOI: 10.1039/d2bm01964g] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nitric oxide (NO) plays a pivotal role in the wound healing process and promotes the generation of healthy endothelium. In this work, a simple method has been developed for fabricating a diselenide grafted gelatin gel, which reduces NO donors such as S-nitroso-N-acetylpenicillamine (SNAP) by glutathione peroxidase-like mechanism to produce NO. Briefly, the process involved covalently conjugating 3,3'-diselenodipropionic acid (DSePA) with gelatin via carbodiimide coupling. The resulting gelatin-DSePA conjugate (G-Se-Se-G) demonstrated NO production upon incubation with SNAP and glutathione (GSH) with the flux of 4.8 ± 0.6 nmol cm-2 min-1 and 1.6 ± 0.1 nmol cm-2 min-1 at 10 min and 40 min, respectively. The G-Se-Se-G recovered even after 5 days of incubation with the reaction mixture retaining catalytic activity up to 74%. Subsequently, G-Se-Se-G was suspended (5% w/v) in water with lecithin (6% w/w of gelatin) and F127 (3% w/w of gelatin) to prepare gel through temperature dependant gelation method. The fabricated G-Se-Se-G gel exhibited desirable rheological characteristics and excellent mechanical stability under storage conditions and did not cause any significant toxicity in normal human keratinocytes (HaCaT) and fibroblast cells (WI38) up to 50 μg ml-1 of selenium equivalent. Finally, mice studies confirmed that topically applied G-Se-Se-G gel and SNAP promoted faster epithelization and collagen deposition at the wound site. In conclusion, the development of a biomimetic NO generating gel with sustained activity and biocompatibility was achieved.
Collapse
Affiliation(s)
- Ram P Das
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - Beena G Singh
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - J Aishwarya
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India.,Advanced Centre for Treatment, Research and Education in Cancer, Mumbai-410210, India
| | - Liladhar B Kumbhare
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India
| | - Amit Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai-400085, India. .,Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| |
Collapse
|
48
|
Shan J, Zhang X, Cheng Y, Song C, Chen G, Gu Z, Zhao Y. Glucose metabolism-inspired catalytic patches for NIR-II phototherapy of diabetic wound infection. Acta Biomater 2023; 157:200-209. [PMID: 36494009 DOI: 10.1016/j.actbio.2022.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Medical patches hold great prospects for diabetic wound administration, while their practical effects in diabetic wound management remain mired by the complexity of wound microenvironments. Here, inspired by the biological processes of glucose metabolism, we present a catalytic microneedle patch that encapsulates near-infrared-II responsive and dual-nanozyme active Au-Cu2MoS4 nanosheets (Au-CMS NSs) for treating diabetic wound infection. Since microneedle patches have great tissue penetration ability, the Au-CMS NSs can be delivered to deep tissues and fully interact with wound environments. Benefitting from the dual nanozyme activities (glucose oxidase and catalase) and near-infrared-II photothermal performances of Au-CMS NSs, the composited catalytic patch realizes in situ glucose consumption, oxygen generation, and bacterial elimination. Notably, their repeatability of near-infrared-II responsive antibacterial capability has been proved both in vitro and in diabetic mice against methicillin-resistant Staphylococcus aureus. The catalytic patch can find wide catalytic applications in wound care and infection prevention. STATEMENT OF SIGNIFICANCE: Effective treatment of diabetic wound infection remains still challenging in the clinic owing to the complex wound microenvironments. Herein, inspired by the biological processes of glucose metabolism in lives, we propose a novel strategy to treat wound infections by modulating the diabetic wound microenvironments. A near-infrared-II (NIR-II) responsive biocatalytic microneedle patch with both glucose oxidase- and catalase-like activities capable of killing bacteria, reducing glucose level, and supplying O2 is developed. The patch not only achieves efficient antibacterial outcomes in vitro, but also is a valuable wound patch for efficient treatment of MRSA-infected wounds in diabetic mice. We anticipate that this therapeutic strategy will provide the applications in chronic inflammation and infections.
Collapse
Affiliation(s)
- Jingyang Shan
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Department of Neurology, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518000, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Xiaoxuan Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yi Cheng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Chuanhui Song
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Guopu Chen
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zhuxiao Gu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China.
| |
Collapse
|
49
|
Ferdinandy P, Andreadou I, Baxter GF, Bøtker HE, Davidson SM, Dobrev D, Gersh BJ, Heusch G, Lecour S, Ruiz-Meana M, Zuurbier CJ, Hausenloy DJ, Schulz R. Interaction of Cardiovascular Nonmodifiable Risk Factors, Comorbidities and Comedications With Ischemia/Reperfusion Injury and Cardioprotection by Pharmacological Treatments and Ischemic Conditioning. Pharmacol Rev 2023; 75:159-216. [PMID: 36753049 PMCID: PMC9832381 DOI: 10.1124/pharmrev.121.000348] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 08/07/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022] Open
Abstract
Preconditioning, postconditioning, and remote conditioning of the myocardium enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and the potential to provide novel therapeutic paradigms for cardioprotection. While many signaling pathways leading to endogenous cardioprotection have been elucidated in experimental studies over the past 30 years, no cardioprotective drug is on the market yet for that indication. One likely major reason for this failure to translate cardioprotection into patient benefit is the lack of rigorous and systematic preclinical evaluation of promising cardioprotective therapies prior to their clinical evaluation, since ischemic heart disease in humans is a complex disorder caused by or associated with cardiovascular risk factors and comorbidities. These risk factors and comorbidities induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury and responses to cardioprotective interventions. Moreover, some of the medications used to treat these comorbidities may impact on cardioprotection by again modifying cellular signaling pathways. The aim of this article is to review the recent evidence that cardiovascular risk factors as well as comorbidities and their medications may modify the response to cardioprotective interventions. We emphasize the critical need for taking into account the presence of cardiovascular risk factors as well as comorbidities and their concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple comorbidities. SIGNIFICANCE STATEMENT: Ischemic heart disease is a major cause of mortality; however, there are still no cardioprotective drugs on the market. Most studies on cardioprotection have been undertaken in animal models of ischemia/reperfusion in the absence of comorbidities; however, ischemic heart disease develops with other systemic disorders (e.g., hypertension, hyperlipidemia, diabetes, atherosclerosis). Here we focus on the preclinical and clinical evidence showing how these comorbidities and their routine medications affect ischemia/reperfusion injury and interfere with cardioprotective strategies.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Ioanna Andreadou
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Hans Erik Bøtker
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sean M Davidson
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Dobromir Dobrev
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Bernard J Gersh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Sandrine Lecour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Marisol Ruiz-Meana
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Coert J Zuurbier
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Pharmahungary Group, Szeged, Hungary (P.F.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece (I.A.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK (G.F.B.); Department of Cardiology, Aarhus University Hospital, Aarhus N, Denmark (H.E.B.); The Hatter Cardiovascular Institute, University College London, London, UK (S.M.D.); Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany (D.D.); Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada (D.D.); Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas (D.D.); Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota (B.J.G.); Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany (G.H.); Cape Heart Institute and Hatter Institute for Cardiovascular Research in Africa, Department of Medicine, University of Cape Town, Cape Town, South Africa (S.L.); Cardiovascular Diseases Research Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Spain (M.R-M.); Laboratory of Experimental Intensive Care Anesthesiology, Department Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands (C.J.Z.); Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.); National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.); Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.); Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
50
|
Zhou L, Sun J, Yang T, Wang S, Shan T, Gu L, Chen J, Wei T, Zhao D, Du C, Bao Y, Wang H, Lu X, Sun H, Lv M, Yang D, Wang L. Improved methodology for efficient establishment of the myocardial ischemia-reperfusion model in pigs through the median thoracic incision. J Biomed Res 2023:1-11. [PMID: 37088562 PMCID: PMC10387751 DOI: 10.7555/jbr.36.20220189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
To investigate the feasibility and effectiveness of establishing porcine ischemia-reperfusion models by ligating the left anterior descending (LAD) coronary artery, we first randomly divided 16 male Bama pigs into a sham group and a model group. After anesthesia, we separated the arteries and veins. Subsequently, we rapidly located the LAD coronary artery at the beginning of its first diagonal branch through a mid-chest incision. Then, we loosened and released the ligation line after five minutes of pre-occlusion. Finally, we ligated the LAD coronary artery in situ two minutes later and loosened the ligature 60 min after ischemia. Compared with the sham group, electrocardiogram showed multiple continuous lead ST-segment elevations, and ultrasound cardiogram showed significantly lower ejection fraction and left ventricular fractional shortening at one hour and seven days post-operation in the model group. Twenty-four hours after the operation, cardiac troponin T and creatine kinase-MB isoenzyme levels significantly increased in the model group, compared with the sham group. Hematoxylin and eosin staining showed the presence of many inflammatory cells infiltrating the interstitium of the myocardium in the model group but not in the sham group. Masson staining revealed a significant increase in infarct size in the ischemia/reperfusion group. All eight pigs in the model group recovered with normal sinus heart rates, and the survival rate was 100%. In conclusion, the method can provide an accurate and stable large animal model for preclinical research on ischemia/reperfusion with a high success rate and homogeneity of the myocardial infarction area.
Collapse
Affiliation(s)
- Liuhua Zhou
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jiateng Sun
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tongtong Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Sibo Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tiankai Shan
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Lingfeng Gu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jiawen Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Tianwen Wei
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Di Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Chong Du
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yulin Bao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Hao Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiaohu Lu
- Department of Cardiac surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Haoliang Sun
- Department of Cardiac surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Meng Lv
- Medical Experimental Animal Center, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Di Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Liansheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|